throbber
J. vet. Pharmacol. Therap. 27, 455-466, 2004.
`
`Bioavailability and its assessment
`
`REVIEW
`
`P. L. TOUTAIN &
`
`A. BOUSQUET-MELOU
`
`UMR 181 Physiopathologie et
`Toxicologie Expérimentales INRA/ENVT,
`Ecole Nationale Vétérinaire de Toulouse,
`Toulouse cedex 03, France
`
`Toutain, P. L., Bousquet-Mélou, A. Bioavailability and its assessment. J. vet.
`Pharmacol. Therap. 27, 455-466.
`
`Bioavailability is a key pharmacokinetic parameter which expresses the
`proportion of a drug administered by any nonvascular route that gains access
`to the systemic circulation. Presented in this review are the different approaches
`to measurementof bioavailability (absolute and relative), including the case in
`which intravenous administration is impossible. The rate of drug absorption is
`also discussed with special emphasis on the possible difficulties encountered
`using Cyax and Tmax or curvefitting to evaluate the rate of drug absorption.
`
`P.L. Toutain, UMR 181 Physiopathologie et Toxicologie Expérimentales INRA/
`ENVT, Ecole Nationale Vétérinaire de Toulouse, 23, chemin des Capelles, 31076
`Toulouse cedex 03, France. E-mail: pl.toutain@envt.fr
`
`To become available in the general circulation, drug should
`gain access to arterial blood. However, as arterial (aortic) blood
`is seldom sampled, bioavailability is normally referred to the
`usualsite of measurement (venous blood). There are occasions
`when drugs delivered to venous blood are not systemically
`available due to an extensive pulmonary first-pass effect (vide
`infra).
`
`ABSORPTION VS. BIOAVAILABILITY
`
`INTRODUCTION
`
`Bioavailability (denoted as F and generally expressed as a
`percentage, F%) quantifies the proportion of a drug which is
`absorbed and available to produce systemic effects. Bioavailabil-
`ity is a fundamental property of a pharmaceutical product for a
`given route of administration. It should be known and shownto
`be reproducible for all drug products intended to produce a
`systemic effect. Bioavailability assessment may also be of value
`for substanceslocally administered and intended to produce only
`local effect,
`in order to demonstrate the absence of systemic
`exposure and to support claims regarding the absence of
`systemic effect, or possible residues in edible tissues of food
`producing species.
`This review will focus on the bioavailability assessment (rate
`and extent) and not on the numerousphysicochemical, physio-
`logical and pathological
`factors capable of
`influencing the
`bioavailability of a drug (see Baggot, 2001). The review by
`Cutler (1981) on the several approaches to compute bioavail-
`ability is still authoritative.
`
`In a physiological context, the terms absorption and bioavaila-
`bility are neither synonymous nor
`interchangeable (Chiou,
`2001). Absorption is only one of the steps separating drug
`administration from its delivery to the site of action. From a
`mechanistic point of view,it can be helpful to distinguish the two
`concepts in order to explain the origin of low bioavailability; for
`example, a drug can be 100% absorbed from a given formulation
`(therefore no possible improvement) but have nevertheless a low
`bioavailability due to breakdown after absorption. This is the
`case for prostaglandin (PgF2«), which undergoes a 90% lung
`first-pass effect (Bonnin et al., 1999) (see review on clearance in
`this issue) and for many drugs undergoingvariable hepatic first-
`pass effect after oral or
`intra-peritoneal administration,
`for
`example propranolol in dog (Bai et al., 1985). To ascribe low
`According to the European Medicines Evaluation Agency (EMEA,
`bioavailability to poor absorption (and expecting to improveit
`humanguidelines) ‘bioavailability means the rate and extent to
`
`which the active substance or active moiety is absorbed from a with a new formulation) when the causeis actuallyafirst-pass
`pharmaceutical form, and becomesavailable at the site of action’
`effect (and not amendable to improvement) can be counterpro-
`(Anonymous, 2001). As the site of action may not be well
`ductive during drug development.
`defined, it is also stated that ‘bioavailability is understood to be
`In the context of bioavailability measurement (not interpret-
`the extent and the rate at which a substanceorits active moiety
`ation), the termsof absorption and bioavailability are often used
`is delivered from a pharmaceutical form, and becomesavailable
`interchangeably (e.g.
`in Gibaldi & Perrier, 1982; Rowland &
`in the generalcirculation’.
`Tozer, 1995) despite the above considerations. Unless stated
`
`DEFINITION OF BIOAVAILABILITY
`
`© 2004 Blackwell Publishing Ltd
`
`CELGENE 2122
`APOTEX v. CELGENE
`IPR2023-00512
`
`455
`
`

`

`456 P.L. Toutain & A. Bousquet-Mélou
`
`otherwise, the word absorption in this review should also be
`understood to be synonymouswith bioavailability.
`
`ABSOLUTE VS. RELATIVE BIOAVAILABILITY
`
`Absolute bioavailability is
`the actual percentage of
`the
`administered dose (from 0 to 100%), which reaches the general
`circulation. Estimation involves comparing drug exposure fol-
`lowing extravascular (e.v.) administration of the tested dosage
`form with that of an intravenous administration (i.v.), assumed
`to be 100% available.
`
`Relative bioavailability involves comparison of two formula-
`tions (or two routes of administration of the same formulation)
`without
`reference to an i.v. administration.
`It should be
`
`emphasized that interpretation of a relative bioavailability trial
`can be of limited value if the absolute bioavailability of the
`reference formulation is not known. Indeed, improving by 100%
`the bioavailability of a given reference formulation (e.g. either by
`manipulating the food regimenin the caseof oral administration
`or modifying the formulation) has different meanings if the
`reference formulation is 5 or 50% bioavailable. In the former
`
`case, the improvementlacks interest, whereas in thelatter it is
`very significant!
`
`BIOAVAILABILITY VS. BIOEQUIVALENCE
`
`Althoughbioavailability is used as an endpoint in bioequivalence
`trials, bioavailability and bioequivalence trials are conceptually
`different. A bioequivalencetrial aims to establish the therapeutic
`equivalence of two formulations (or two routes of administra-
`tion). It is not concerned with documenting the physiological
`factors capable of influencing systemic exposureof the drug. In a
`bioequivalence trial, animals are only biological
`test-tubes,
`required for an in vivo quality control for different formulations
`(Toutain & Koritz, 1997). In contrast, in a bioavailability trial, it
`is the animal physiology and possibly pathology (age, sex, food
`intake, disease state and severity, etc.) which are of primary
`interest.
`
`IT IS A MISCONCEPTION THAT A LOW
`BIOAVAILABILITY CAN ALWAYS BE COMPENSATED BY
`INCREASING THE RECOMMENDED DOSE
`
`It is frequently stated that ‘the absolute bioavailability of this
`drug/product is of no consequence (e.g. a pour-on for cattle),
`because the dose has been sufficiently increased to guarantee
`clinicalefficacy’. This statement is questionable in general terms,
`as it is a primary objective of all rational drug development
`programmes to market drug products having the highest
`possible systemic bioavailability.
`More specially, a low bioavailability can be a major source of
`therapeutic variability. If the mean bioavailability in a group of
`animals is 100%,
`there is no possibility of inter-individual
`
`variability in drug exposure because of the different factors
`influencing the bioavailability. In contrast, when the mean
`bioavailability is
`low (e.g.
`10%), a large inter-individual
`variability will be expected, with some subjects having a very
`low (e.g. 5%), and with others having a higher bioavailability
`(e.g. 20%), thus leading to exposures varying from 1 to 4,i.e. by
`400% (and consequently, a lack of reproducibility of
`this
`formulation in termsofclinical efficacy).
`Fig.
`1 showstherelationship between absolute bioavailability
`and inter-individual variability for a set of drugs in man
`(Hellriegel et al., 1996). In veterinary medicine, similar obser-
`vations can be made, e.g.
`in the horse, the bioavailability of
`rifampicin when the drug is administered 1 h before feeding is
`68 + 26% (coefficient of variation = 38%), whereas when the
`same drug is administered 1 h after feeding, the bioavailability is
`26 + 17%, ie. with a coefficient of variation of 67% (Baggot,
`2001). In pigs, the mean absolute bioavailability of chlortetra-
`cycline administered by the oral route in fasted animals is low
`(19%) andvariable, ranging from 9 to 30% (Kilroyet al., 1990),
`which is not satisfactory in terms of
`the prudent use of
`antibiotics. Indeed, the emergenceof resistance is often because
`of an underexposure of small animal population subgroups
`despite an appropriate average dose.
`Wheneverbioavailability is low, drug companies may attempt
`to increase the dose to achieve an appropriate drug exposure.
`However,
`it may not be recognized that the dose is generally
`increased more than proportionally to the mean bioavailability
`factor in order to ensure drug efficacy in animals having the
`lowest bioavailability. For instance,
`for a hypothetical drug
`having a mean bioavailability of 33% (with some subjects
`having 20% bioavailability, and others 50%) and for which an
`
`100
`
`125
`
`CV(%)
`
`Fig. 1. Relationship between bioavailability (F%) and inter-subject
`variability (CV) in man (Hellriegel et al., 1996). Data correspond to 100
`different drugs.
`
`© 2004 Blackwell Publishing Ltd,J. vet. Pharmacol. Therap. 27, 455-466
`
`

`

`exposure corresponding to 100% bioavailability is required, the
`dose should be multiplied by 5 (not by 3) to guarantee that the
`Not only extent, but also rate of absorption needs to be known,
`subjects with the lowest bioavailability are fully exposed. By
`because both determine the shape of the plasma concentration
`doing this, the subjects having aninitial bioavailability of 50%
`vs. time curve and may influence the drug effect (e.g. concen-
`are unnecessarily overexposed by a factor of 2.5! If such an
`tration vs. time dependent antibiotic, duration of protection for
`overexposure is considered to be detrimental on safety or other
`an avermectin, etc.). Similarly, the toxic- or side-effect can be
`grounds,
`the dose can only be doubled. Then,
`it will be the
`markedly different when absorption rates for new formulations
`subjects with the poorest availability who will be underexposed,
`differ widely (Fig. 3). It is also essential to know the rate of
`with a possible reduction of clinical efficacy or worse, by creating
`availability when a drug is presented as a specific formulation,
`a situation favouring the emergenceofresistance (antibiotic and
`antiparasitic drugs). Finally, for a drug having both a narrow
`intended to precisely control
`the rate of drug delivery (e.g.
`
`therapeutic window andapoorbioavailability, it is possible to modified released products such as rumen retention device,
`encounter a situation for which no dose is able to expose
`vaginal sponge,etc.).
`adequately all the animals within a population (Fig. 2).
`A poor(oral) bioavailability is also a risk factor for possible
`interaction. Indeed,for a drug havinga low bioavailability, there is
`room for increasing exposure anda possibility of leading to over-
`exposure, as exemplified in man with felodipine. Felodipine is an
`anti-arrhythmic drug which has a low (approximately 15%) and
`erratic bioavailability becauseof a gutfirst-pass effect (metabolism
`by intestinal CYP3A4). Ingestion of grapefruit (which inhibits
`intestinal CYP450) can greatly increase the systemic exposure to
`felodipine (from 1 to 12 times between individuals) (Bailey et al.,
`2000). In veterinary medicine, oral bioavailability of endectocides
`is relatively low,
`leading to a possible interaction with food
`components as shown between moxidectin and quercetin, a
`naturalflavonoid occurring in plants, and whichis a modulatorof
`P-glycoprotein (Dupuyet al., 2003).
`Finally,
`in order to document exposure variability and to
`anticipate possible under or over exposure, the measurementof
`absolute bioavailability is mandatory for any new drug formu-
`lation. Thus,
`that no serious drug development should be
`performed without intravenous data information.
`
`RATE OF BIOAVAILABILITY
`
`Bioavailability and its assessment 457
`
`ABSOLUTE BIOAVAILABILITY BY THE I.V. ROUTE IS
`NOT ALWAYS 100%
`
`By assumption, a drug administered by the i.v. route has 100%
`bioavailability. This is true only if the active substance reaches
`arterial blood without loss. Drugs are generally administered by
`the iv. route and havefirst to cross the pulmonary circulation
`before gaining access to arterial blood. Lungs can be the site of
`an extensivefirst-pass effect and reduce drug availability. This is
`the case for prostaglandins or some amines(see our companion
`paper on plasmaclearance in this issue, Toutain & Bousquet-
`Mélou 2004, pp. 415-425).
`After the i.v. administration of a pro-drug, bioavailability can
`be <100%. For example, methylprednisolone(a corticosteroid) is
`
`Concentrations(mg/L)
`
`AUC
`
`
`
`
`
`
`
`
`
`Overexposure of some
`animals (side effects)
`
`
`
`Undesired
`exposure
`
`herapeutic
`exposure
`
`
`
`Underexposure
`of some animals
`therapeutic failure, resistance)
`
`Doses
`
`1
`
`3
`
`2
`
`Fig. 2. Low bioavailability and the impossibility of establishing a safe and
`efficacious dosage regimenfor all animals. The figure shows, diagram-
`matically, the circumstance for which a drug formulation having a low
`and variable bioavailability cannot be administered at the same
`efficacious dosage in all animals dueto a relatively narrow therapeutic
`window. With dose 1, all animals fail to achieve effective therapeutic
`exposure. Increasing the dose threefold guarantees that all animals now
`have an exposure abovethe therapeutic threshold, but animals with the
`highest bioavailability are now above the undesired threshold. Finally,
`increasing the dose by twofold guarantees that no animal undergoes
`undesired exposure, but the animals having the lowest bioavailability are
`under-exposed.
`
`© 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 455-466
`
`
`
`Undesired concentrations
`
`Therapeutic concentrations
`
`0
`
`6
`
`12
`
`18
`
`24
`
`Time(h)
`
`Fig. 3. Drug effect and the rate of drug absorption. For three formula-
`tions (A, B and C) having the same bioavailability (same AUC), effect
`(therapeutic and undesired) differs depending on rate of absorption. For
`formulation A, the rate constant of absorption is high and the peak
`plasma concentration is above the safe concentration. In contrast, for
`formulation C, the rate of absorption is too low to allow plasma
`concentration to reach effective plasma concentrations(e.g. for a time-
`dependentantibiotic). Only formulation B gives a plasma concentration
`profile within the therapeutic window.
`
`

`

`458 P.L. Toutain & A. Bousquet-Mélou
`
`a poorly hydrosoluble drug. In order to administer it by thei.v.
`route, a hydrosoluble ester has been synthesized (methylpredni-
`solone sodium succinate or Solumedrol®). This is a pro-drug of
`methylprednisolone, and the ester must be hydrolysed to release
`its active moiety (i.e. methylprednisolone). In the dog, it was
`shownthat thei.v. bioavailability of methylprednisolone from its
`succinic ester was only 40% (Toutain et al., 1986) (Fig. 4). Other
`examples of pro-drugs are inactive esters of macrolides, antibi-
`otics such as pivampicillin and chloramphenicol succinate and
`antiviral pro-drugs.
`
`THE MEASUREMENT OF ABSOLUTE BIOAVAILABILITY
`
`There are many methods used to evaluate the extent of systemic
`availability,
`the most classical one consisting of comparing
`plasma exposure (AUC) after an i.v. and an e.v. administration.
`Classically, the bioavailability factor is obtained by the following
`equation:
`
`P%
`
`Dose;y.
`_ AUCex.
`~ AUGiy,
`Dosegy.
`
`x 100.
`
`(1)
`
`In Eqn 1, AUCis the area underthe plasma(total or free) drug
`concentration or total blood concentration-time curve and
`
`Dosej;,,, and Dose,,, are the doses actually administered to
`evaluate F%.
`
`There is a compelling assumption underlying Eqn 1: the i.v.
`and the e.v. clearances must be equal. Indeed, according to the
`mass balance consideration, what is actually measured when
`using Eqn1, is the ratio of the bioavailable doses after an e.v. and
`an i.v. administration; the bioavailable dose actually corresponds
`to the dose eliminated from plasma, as given by the general
`relationship:
`
`Dose = Body clearance x AUC.
`
`Equation 1 is the reduced form of the following equation:
`
`_ AUCwy, X Cley, x Dosejy,
`Ph = AUC. X Clix. X Dosegy.
`
`(2)
`
`(3)
`
`if the body
`and Eqn 3 reduces to Eqn 1 if Cl, = Cl.y., ie.
`clearance is a dose and time invariable parameter(linear phar-
`macokinetics).
`Generally, bioavailability is measured experimentally using a
`crossover study design andit is necessary to check for a possible
`carry-overeffect, i.e. a differential residual effect of the first period
`over the second one. For example, hepatic enzymatic inhibition
`(or induction) after the first administration can modify the
`clearance during the second period. Therefore,
`the washout
`period should be long enough, not only to guarantee the absence
`of residual drug plasma concentration butalso the lack of residual
`drug effect on clearance (induction/inhibition). This nonlinearity
`due to time dependencyis also called nonstationarity.
`The presenceofa significant (differential) carry-over effect can
`be detected by properly analysing the design (by testing the
`sequence effect), but if an unbalanced design has been carried
`out(i.e. by performingi.v. route of administration for all animals
`in thefirst period, and e.v. in the secondperiod), an equal carry-
`over will be obtained, which can betotally confounded with a
`period effect
`leading to a possibly large overestimation (or
`underestimation) of bioavailability. With this type of design, a
`bioavailability higher than 100% can be computed.
`For random inter-occasion clearance variability, it has been
`suggested to correct the computed bioavailability factor by the
`terminal half-life (Wagner, 1967) using the following equation:
`
`_ AUCex.
`(1/2, iv.
`~ AUGy,
`1/2, ev.
`
`x 100.
`
`(4)
`
`F%
`
`105
`
`104
`
`The logic for the so-called half-life correction is rooted in the
`relationship between t,/2 and clearance from the following
`equation:
`
`hp=
`
`0.693 x Vq
`Clearance °
`
`(5)
`
`¢MPafter IV administration of MP Plasmaconcentration(ng/mL) 102\we after administration of MPS
`
`
`
`103
`
`Assuming that Vg is invariable, tj/2 can be incorporated as a
`surrogate of clearance in Eqn 3.
`This correction should be used cautiously and accepted only
`if it results in a substantial decrease in the variability of the
`MPS
`10
`Lop|ff
`results, or in order to avoid a meanbioavailability higher than
`0
`60 120
`240
`360
`480 min
`100%. Indeed, the estimation of terminal half-life (contrary to
`body clearance) is not very robust. In addition, if the terminal
`half-life does not represent
`the drug elimination but rather
`drug absorption (flip-flop), the correction becomestotallyillicit
`and its unjustified use may lead to markedly underestimated
`F%. For the same objective, an equation correction can be
`made using renal clearance, which could be independently
`evaluated during a bioavailability trial. According to Karlsson
`and Sheiner (1994),
`the best way to handle random inter-
`occasion clearance variability is to analyse all
`the subjects
`simultaneously with a nonlinear mixed effect model.
`
`Fig. 4. Absolute bioavailability of methylprednisolone in the dog.
`Methylprednisolone (MP) is a nonhydrosoluble steroid which can be
`administered by the i.v. route using a hydrosoluble salt of a succinate
`ester (methylprednisolone sodium succinate, MPS). After i.v. adminis-
`tration of MPS,its concentration decreases rapidly providing the active
`moiety, i.e. MP. Using this MP plasma concentration profile to estimate
`the MP bioavailability of another formulation would lead to gross
`overestimation of the true MP bioavailability, because only 44% of the
`administered MPS wastransformed into MP. Thetruebioavailability of
`an MP formulation should be obtained by administering MP itself via the
`i.v. route (Toutain et al., 1986).
`
`© 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 455-466
`
`

`

`MEASUREMENT OF ABSOLUTE BIOAVAILABILITY
`WHEN PLASMA DRUG CONCENTRATIONS ARE NOT
`DIRECTLY MEASURABLE
`
`EVALUATION OF BIOAVAILABILITY WHEN TERMINAL
`HALF-LIFE IS VERY PROLONGED AND LIMITS THE USE
`OF A CROSS-OVER DESIGN
`
`Bioavailability and its assessment 459
`
`In someinstances, the measurementof plasma concentration of
`the administered drugis impossible, either because an appropriate
`analytical technique is not available, or more often because the
`drug is rapidly transformed into an active metabolite (e.g. 4-
`methylaminoantipyrine or 4-MMA from dipyrone). In these
`conditions, the drug absolute bioavailability can be evaluated by
`measuring the AUCof its metabolite using the following equation:
`
`F%
`
`AUC metabolite
`e.v., parent drug
`_
`~~ AUC.metabolite
`i.v., parent drug
`
`x 100.
`
`(6)
`
`For some drugs having a long terminal half-life (e.g. avermec-
`tins, moxidectin, salicylanilides, etc.) the absolute bioavailability
`is seldom measured, becausetrials involving two (long) periods
`separated by a washout period of 10 times the terminal half-life
`are considered as prohibitive. In addition, nothing guarantees
`the invariance of the plasma clearance over such a prolonged
`period of time, especially in growing animals,
`thus making
`invalid the use of Eqn 1.
`the
`to consider
`A possible solution to this problem is
`estimation of bioavailability by a semi-simultaneous drug
`administration such as
`that described by Karlsson and
`The condition for using Eqn 6 is that the metabolite must not
`Bredberg (1990). The principle of
`this method comprises
`be formed at the administration site or byafirst-pass effect. In
`administering one of the two doses(i.v. then e.v., or e.v. then
`the same circumstance (nofirst-pass effect) a nonspecific assay
`iv.) at an optimal
`interval and fitting simultaneously the
`(e.g. radioactivity), measuring both the drug and its metabo-
`entire curve obtained with an appropriate model,
`including
`lite(s), can be used to determine bioavailability. However, a
`and estimating the rate constant of absorption,
`lag, and the
`nonspecific assay cannot be used for nonlinear systems.
`bioavailability factor.
`If a drug is metabolized solely by the liver and subjected to a
`It was shownthat the precision of the method wasinfluenced
`significant hepatic first-pass effect, then Eqn 1 will be appropriate
`by the doserate, the order of administration, the e.v. vs. i.v. dose
`to measure the absolute bioavailability of an oral formulation,
`ratio, the duration of the sampling, and the interval between the
`whereasEqn 6 will give the fraction of the dose actually absorbed
`doses. This approach deserves to be encouraged in veterinary
`after the oral administration (for more information see Weiss,
`medicine as a screening method, when intra-animalvariability is
`1990).
`expected (e.g.
`time-dependent variation of clearance during
`growth), or to reduce the total numberof blood samplings. The
`most appropriate design can be determined by Monte Carlo
`simulations. The relative bioavailability of two e.v. formulations
`can also be documented using this approach.
`
`MEASUREMENTOF BIOAVAILABILITY USING URINARY
`CONCENTRATIONS
`
`Absolute bioavailability can be assessed by measuring the
`amountof drug excreted in urine (or any other biological fluid
`or excreta) using the following equation:
`‘CO
`
`U,
`€.V.
`Dose;y.
`OO
`xXx iv,
` DOSe€ey,
`
`x 100,
`
`(7)
`
`F% =
`
`where X™ is the total amount of drug eliminated in urine (or
`other biologicalfluid).
`The assumption underlying Eqn 7 is that the ratio of renal
`clearance andtotal clearance is the samefor the i.v. and e.v.
`
`administrations. The main limit of the urinary approachis the
`need to collect urine (or faeces, milk, etc.) until almost all the
`drug has been excreted. The use of partial urine collection is
`theoretically possible but
`requires several assumptions not
`always easy to check. Urinary metabolite, provided that it is
`not formed by a first-pass effect, can be used for absolute
`bioavailability measurement:
`co, metabolite
`U,e.v., parent drug
`x® metabolite
`u, i.v., parent drug
`
`Dose;y.
`Doseg.y.
`
`x 100.
`
`(8)
`
`F% =
`
`Equation 8 can be usedto assess therelative bioavailability of
`two formulations administered by the sameroute regardless of
`the presence ofa first-pass effect.
`
`© 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 455-466
`
`MEASUREMENT OF AN ABSOLUTE BIOAVAILABILITY
`WHEN AN I.V. ADMINISTRATION IS NOT POSSIBLE
`
`Sometimes, it is difficult or impossible to administer a drug by
`the i.v.
`route, but an indirect evaluation of
`the absolute
`bioavailability is
`still possible if a fraction of
`the dose is
`eliminated by the kidney (or any other accessible body fluid),
`andif there is a sufficiently large variability in renal clearance
`amongthedifferent subjects under investigation (Hinderling &
`Shi, 1995).
`The principle of this method is as follows; after an i.v.
`administration:
`
`
`Dose
`
`AUCG,y,
`
`= Clot = Clr + Chr,
`
`(9)
`
`is the body (plasma) clearance, Clg is the renal
`where Clio,
`clearance and Clppg,
`the non-renal clearance. After an e.v.
`administration, the following relationship holds:
`
`Dose — Clot 1
`AUCex. FF F
`
`(10)
`
`where F is the bioavailability factor to be estimated. Assuming
`that Clyp/F is a constant and Clg is a variable, Eqn 10 is the
`
`

`

`WHY MAYA BIOAVAILABILITY HIGHER THAN 100%
`BE COMPUTED?
`
`Bioavailabilities higher than 100% are regularly reported, which
`is conceptually impossible. The reasons for this are numerous,
`including experimental errors and nonfulfilment of the assump-
`tion for computation of absolute bioavailability (Martinez,
`1998). Table 1 gives the main reasons for obtaining a bioavail-
`ability higher than 100%.
`
`RELATIVE BIOAVAILABILITY BETWEEN TWO
`FORMULATIONS OR TWO E.V. ROUTES OF
`ADMINISTRATION
`
`Relative bioavailability can be measured by comparing the AUC
`of the two tested formulations at the same dose levels using the
`following equation:
`
`AUCeest
`F% =——_———_
`AUCreference x 100.

`
`(13)
`
`Relative bioavailability can also be evaluated under steady-
`state conditions (Fig. 6), because the total AUC over a dosing
`interval at steady-state (i.e. AUC,) is equal to the total AUCy_..
`after a single dose administration.
`Therefore, under steady-state conditions (obtained for exam-
`ple with formulation A), AUC,,, is measured; then formulation
`B is
`immediately administered (i.e. without any washout
`period), and when a new steady-state is obtained with
`formulation B (i.e. after a delay of 4-5 times the terminal
`half-life), AUC,
`is then measured. This method is useful for
`drugs having a long terminal half-life, and for which a
`conventional cross-over design cannot be extended over several
`months because of the requirement for a washout period of
`approximately 10 times the terminal half-life. Another advant-
`age of this method is that fewer data points are required to
`characterize the AUC over the dosing interval as the time
`course of drug concentration at equilibrium is more stable than
`after a single dose administration. The condition required to use
`this method is that all
`the bioavailable amounts of the two
`
`tested formulations have been absorbed during the dosing
`interval, i.e. that absorption does not continue after the end of
`the dosage interval. Urinary excretion data can be used in the
`same way. Urinary excretion data can be used at steady-state
`using the following equation:
`
`Xssu, test
`F% == x
`u, reference
`
`100,
`
`(14)
`
`where XS denotes the amountof drug excreted in the urine over
`a dosing interval at steady-state. Equation 14 is used for the
`same dosage regimen for both formulations. The advantage of
`this approach is the duration of the collecting period (corres-
`pondingto the dosage interval), which can be muchshorter than
`after a single dose administration. The condition to apply Eqn 14
`is to have reached an initial steady-state with the reference
`
`© 2004 Blackwell Publishing Ltd,J. vet. Pharmacol. Therap. 27, 455-466
`
`460 P. L. Toutain & A. Bousquet-Mélou
`
`(y=mx+c) which can be
`line
`equation of a straight
`visualized by plotting Dose/AUC,,,, against Cla; the slope is 1/
`F and the intercept Clyp/F. These two parameters of the line
`are obtained by curve fitting (Fig. 5). The conditions for this
`approach are twofold: the drug must be cleared mainly by the
`renal route (or any other measurable route), and patients
`under
`investigation should display a large inter-individual
`variability in their renal clearance (see Hinderling, 2003 for
`application of the method).
`If
`it
`is known that a drug is exclusively (or almost
`exclusively) eliminated by an experimentally accessible route
`of elimination (urine, faeces), as is the case for eprinomectin,
`which is almost
`totally eliminated unchanged by faeces in
`cattle, absolute bioavailability can be obtained without
`i.v.
`administration by collecting all
`the effluents (mass balance
`principle). However, for a long acting drug (e.g. avermectins),
`collecting all the faeces can be cumbersome. An alternative
`and less demanding method would involve measuring,
`for
`some limited periods of time (e.g. 24 h), the faecal clearance
`(Cleces) using the following equation:
`Amountexerted in faeces over a given period of time
`=
`Plasma AUC over the sameperiod of time
`
`Clraeces
`faeces
`
`(11)
`
`Simultaneously, the total plasma AUCy_.. should be evaluated
`(this is easier to determine than to collect all the faeces over
`several weeks), and the absolute bioavailability can then be
`computed using the following equation:
`
`F%
`
`_ AUCo~26 x Cltaeces
`Administered dose’
`
`(12)
`
`to the total body
`is assumed to be equal
`Here Clmeces
`clearance, and consequently the quantity eliminated via faeces
`(Cltreces X AUCp_..) is equal to the bioavailable dose.
`
`Dose
`
` 1—
`
`— =slope
`
`LEe
`
`
`AUC.y,
`
`Intercept = ——_ Clyyr
`
`Clr
`
`Fig. 5. Evaluation of absolute bioavailability when intravenous(i.v.)
`administration is not possible. Wheni.v. administration is not possible
`but urine sample collection is possible (or any other matrix as faeces,
`etc.), then absolute bioavailability can be measured. The absolute
`clearance of the excretory pathway should be evaluated andthe inter-
`individualvariability of this absolute clearance should be large enough to
`use a regression approach in order to computebioavailability. Bioavail-
`ability is estimated by the slope of the straight line between the measured
`absolute (renal) clearance and the apparent extra-vascular (Dose/AUC)
`clearance (see text for further explanation).
`
`

`

`Bioavailability and its assessment 461
`
`Table 1. Selected factors leading to a bioavailability higher than 100%
`
`1. Experimental errors during the in-life phase of the experiment
`Dose administered by the e.v. route is too high (grosserror, different salts or esters without correction for the molecular weight, inappropriate scoring
`of tablets etc.)
`Dose administered by i.v. route is too low or not available [gross error, physical interaction with the injecting material (e.g. lidocaine), drug is not
`stable in solution (e.g. peptide during infusion). In vivo precipitation of an extemporaneoussolution, a different salt or ester without correction for the
`molecular weight, incomplete transformation of an ester pro-drug to its active moiety (see Fig. 4)]
`Exchange of drug between animals raised in groups(licking in cattle, coprophagy in dogs,etc.)
`2. Experimental errors during sampling, preparation and conservation of samples
`Sampling in the jugular vein homolateral to an ear-implantin cattle
`Contamination of the extra-vascular samples when working with a pour-on formulation
`Nonstability of the drug in the i.v. samples: photodegradation (carprofen), delay to centrifugation and freezing for longer period comparedwith e.v.
`samples
`Insufficient samples during the initial phase after the i.v. bolus administration or the upswing of the curve after an i.v. infusion (see Fig. 7).
`3. Analytical technique
`For safety reasons, the administered dose using i.v. route is lower than for extra-vascular route, and the LOQ ofthe analytical phase is too high and
`fails to detect a part of the iv. AUC
`Non-enantioselective analytical technique for a racemate having an enantioselective disposition (e.g. possible presystemic chiral inversion in the
`digestive tract favouring the enantiomer having the lowest clearance)
`Enantioselective analytical technique for a racemate having an enantioselective disposition (e.g. presystemic chiral inversion of ibuprofen in the
`digestive tract of rabbit increases the S(+) from the R(—) ibuprofen (Doki et al., 2003)
`Bacteriological analytical technique for antibiotics overestimates bioavailability when a fraction of the drug is metabolized into a more

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket